DOI QR코드

DOI QR Code

Agglomeration of human dermal fibroblasts with ECM mimicking nano-fragments and their effects on proliferation and cell/ECM interactions

  • Received : 2018.05.01
  • Accepted : 2018.06.11
  • Published : 2018.11.25

Abstract

Here, we engineered spheroids by using ECM mimicking nano-fragments (NFs) with fibroblasts and investigated their effect on proliferation and cell/ECM interactions. NF incorporation resulted in formation of a stable spheroid, which improved proliferation and viability of cells by assisting oxygen transport confirmed by LOX-1 staining. In addition, hypoxic and apoptotic genes were significantly downregulated in spheroids with PD-NFs. Furthermore, ECM and cell junction proteins were highly expressed. Overall, our findings suggest that incorporation of NFs within spheroids for assembly with various cell types can be an alternative approach for 3D cell culture in many applications.

Keywords

Acknowledgement

Supported by : National Research Foundation of Korea (NRF)

References

  1. P. Bajaj, R.M. Schweller, A. Khademhosseini, J.L. West, R. Bashir, Annu. Rev. Biomed. Eng. 16 (2014) 247. https://doi.org/10.1146/annurev-bioeng-071813-105155
  2. E. Knight, S. Przyborski, J. Anat. 227 (2015) 746. https://doi.org/10.1111/joa.12257
  3. E. Fennema, N. Rivron, J. Rouwkema, C. van Blitterswijk, J. de Boer, Trends Biotechnol. 31 (2013) 108. https://doi.org/10.1016/j.tibtech.2012.12.003
  4. V. van Duinen, S.J. Trietsch, J. Joore, P. Vulto, T. Hankemeier, Curr. Opin. Biotechnol. 35 (2015) 118. https://doi.org/10.1016/j.copbio.2015.05.002
  5. A. Sachar, T.A. Strom, M.J. Serrano, M.D. Benson, L.A. Opperman, K.K. Svoboda, X. Liu, J. Biomed. Mater. Res. A 100 (2012) 3029.
  6. S. Bosi, R. Rauti, J. Laishram, A. Turco, D. Lonardoni, T. Nieus, M. Prato, D. Scaini, L. Ballerini, Sci. Rep. (Nat. Publ. Group) 5 (2015) 9562.
  7. V. Krishnan, L.A. Shuman, D.M. Sosnoski, R. Dhurjati, E.A. Vogler, A.M. Mastro, J. Cell. Physiol. 226 (2011) 2150. https://doi.org/10.1002/jcp.22550
  8. F. Guilak, D.M. Cohen, B.T. Estes, J.M. Gimble, W. Liedtke, C.S. Chen, Cell Stem Cell 5 (2009) 17. https://doi.org/10.1016/j.stem.2009.06.016
  9. Y.-P. Lo, Y.-S. Liu, M.G. Rimando, J.H.-C. Ho, K.-h. Lin, O.K. Lee, Sci. Rep. 6 (2016) 21253. https://doi.org/10.1038/srep21253
  10. I.A. Janson, A.J. Putnam, J. Biomed. Mater. Res. A 103 (2015) 1246. https://doi.org/10.1002/jbm.a.35254
  11. R. Edmondson, J.J. Broglie, A.F. Adcock, L. Yang, Assay Drug Dev. Technol. 12 (2014) 207. https://doi.org/10.1089/adt.2014.573
  12. D. Antoni, H. Burckel, E. Josset, G. Noel, Int. J. Mol. Sci. 16 (2015) 5517. https://doi.org/10.3390/ijms16035517
  13. S.L. Chia, C.Y. Tay, M.I. Setyawati, D.T. Leong, Small 11 (2015) 702. https://doi.org/10.1002/smll.201401915
  14. C.L. Rettinger, A.B. Fourcaudot, S.J. Hong, T.A. Mustoe, R.G. Hale, K.P. Leung, Cell Tissue Res. 358 (2014) 395. https://doi.org/10.1007/s00441-014-1939-0
  15. V. Mironov, R.P. Visconti, V. Kasyanov, G. Forgacs, C.J. Drake, R.R. Markwald, Biomaterials 30 (2009) 2164. https://doi.org/10.1016/j.biomaterials.2008.12.084
  16. X. Cui, Y. Hartanto, H. Zhang, J. Royal Soc. Interface 14 (2017) 20160877. https://doi.org/10.1098/rsif.2016.0877
  17. K. Mineda, J. Feng, H. Ishimine, H. Takada, S. Kuno, K. Kinoshita, K. Kanayama, H. Kato, T. Mashiko, I. Hashimoto, Stem Cells Transl. Med. 4 (2015) 1511. https://doi.org/10.5966/sctm.2015-0037
  18. M.L. Skiles, S. Sahai, L. Rucker, J.O. Blanchette, Tissue Eng. Part A 19 (2013) 2330. https://doi.org/10.1089/ten.tea.2012.0750
  19. G. Mehta, A.Y. Hsiao, M. Ingram, G.D. Luker, S. Takayama, J. Control. Release 164 (2012) 192. https://doi.org/10.1016/j.jconrel.2012.04.045
  20. Z. Cesarz, K. Tamama, Stem Cells Int. 2016 (2015).
  21. D. Loessner, K.S. Stok, M.P. Lutolf, D.W. Hutmacher, J.A. Clements, S.C. Rizzi, Biomaterials 31 (2010) 8494. https://doi.org/10.1016/j.biomaterials.2010.07.064
  22. F. Gattazzo, A. Urciuolo, P. Bonaldo, Biochim. Biophys. Acta Gen. Subj. 1840 (2014) 2506. https://doi.org/10.1016/j.bbagen.2014.01.010
  23. W. Asghar, R. El Assal, H. Shafiee, S. Pitteri, R. Paulmurugan, U. Demirci, Mater. Today 18 (2015) 539. https://doi.org/10.1016/j.mattod.2015.05.002
  24. S. Breslin, L. O'Driscoll, Drug Discov. Today 18 (2013) 240. https://doi.org/10.1016/j.drudis.2012.10.003
  25. S. Guven, P. Chen, F. Inci, S. Tasoglu, B. Erkmen, U. Demirci, Trends Biotechnol. 33 (2015) 269. https://doi.org/10.1016/j.tibtech.2015.02.003
  26. Y. Wang, L. Zhao, C. Tian, C. Ma, J. Wang, Anal. Methods 7 (2015) 10040. https://doi.org/10.1039/C5AY02466H
  27. Y. Tan, D. Richards, R. Xu, S. Stewart-Clark, S.K. Mani, T.K. Borg, D.R. Menick, B. Tian, Y. Mei, Nano Lett. 15 (2015) 2765. https://doi.org/10.1021/nl502227a
  28. J. Park, Y.S. Kim, S. Ryu, W.S. Kang, S. Park, J. Han, H.C. Jeong, B.H. Hong, Y. Ahn, B.S. Kim, Adv. Funct. Mater. 25 (2015) 2590. https://doi.org/10.1002/adfm.201500365
  29. M. Yamada, A. Hori, S. Sugaya, Y. Yajima, R. Utoh, M. Yamato, M. Seki, Lab Chip 15 (2015) 3941. https://doi.org/10.1039/C5LC00785B
  30. R.L. Carpenedo, A.M. Bratt-Leal, R.A. Marklein, S.A. Seaman, N.J. Bowen, J.F. McDonald, T.C. McDevitt, Biomaterials 30 (2009) 2507. https://doi.org/10.1016/j.biomaterials.2009.01.007
  31. A.M. Bratt-Leal, R.L. Carpenedo, M.D. Ungrin, P.W. Zandstra, T.C. McDevitt, Biomaterials 32 (2011) 48. https://doi.org/10.1016/j.biomaterials.2010.08.113
  32. P.R. Baraniak, M.T. Cooke, R. Saeed, M.A. Kinney, K.M. Fridley, T.C. McDevitt, J. Mech. Behav. Biomed. Mater. 11 (2012) 63. https://doi.org/10.1016/j.jmbbm.2012.02.018
  33. C.C. Ahrens, Z. Dong, W. Li, Acta Biomater. (2017).
  34. Y.M. Shin, W.-G. La, M.S. Lee, H.S. Yang, Y.-M. Lim, J. Mater. Chem. B 3 (2015) 8375. https://doi.org/10.1039/C5TB01310K
  35. T. Ahmad, J. Lee, Y.M. Shin, H.J. Shin, S.K. Madhurakat Perikamana, S.H. Park, S. W. Kim, H. Shin, Acta Biomater. 64 (2017) 161. https://doi.org/10.1016/j.actbio.2017.10.022
  36. Y.M. Shin, T.G. Kim, J.-S. Park, H.-J. Gwon, S.I. Jeong, H. Shin, K.-S. Kim, D. Kim, M.-H. Yoon, Y.-M. Lim, J. Mater. Chem. B 3 (2015) 2732. https://doi.org/10.1039/C3TB21830A
  37. Y.M. Shin, Y.B. Lee, H. Shin, Coll. Surf. B: Biointerfaces 87 (2011) 79. https://doi.org/10.1016/j.colsurfb.2011.05.004
  38. J. Lee, T. Ahmad, M.S. Lee, H. Yang, D.-G. Kim, K. Kim, B. Kwon, H. Shin, Tissue Eng. (2017).
  39. Y. Zhu, Z. Mao, C. Gao, RSC Adv. 3 (2013) 2509. https://doi.org/10.1039/C2RA22358A
  40. T.G. Kim, T.G. Park, Macromol. Rapid Commun. 29 (2008) 1231. https://doi.org/10.1002/marc.200800094
  41. R. Ravichandran, J.R. Venugopal, S. Sundarrajan, S. Mukherjee, S. Ramakrishna, Biomaterials 33 (2012) 846. https://doi.org/10.1016/j.biomaterials.2011.10.030
  42. S. Nirasay, A. Badia, G. Leclair, J.P. Claverie, I. Marcotte, Materials 5 (2012) 2621. https://doi.org/10.3390/ma5122621
  43. R. Luo, L. Tang, S. Zhong, Z. Yang, J. Wang, Y. Weng, Q. Tu, C. Jiang, N. Huang, A.C.S Appl. Mater. Interfaces 5 (2013) 1704. https://doi.org/10.1021/am3027635
  44. S.K. Madhurakkat Perikamana, J. Lee, T. Ahmad, Y. Jeong, D.-G. Kim, K. Kim, H. Shin, A.C.S Appl. Mater. Interfaces 7 (2015) 8798. https://doi.org/10.1021/acsami.5b01340
  45. S.K. Madhurakkat Perikamana, J. Lee, Y.B. Lee, Y.M. Shin, E.J. Lee, A.G. Mikos, H. Shin, Biomacromolecules 16 (2015) 2541. https://doi.org/10.1021/acs.biomac.5b00852
  46. S. Tajima, Y. Tabata, J. Tissue Eng. Regen. Med. 7 (2013) 801.
  47. T.-T. Zhu, W.-F. Zhang, P. Luo, Z.-X. Qian, F. Li, Z. Zhang, C.-P. Hu, Life Sci. 174 (2017) 35. https://doi.org/10.1016/j.lfs.2017.02.016
  48. G. Kagiya, R. Ogawa, F. Hyodo, K. Yamashita, M. Nakamura, A. Ishii, Y. Sejimo, S. Tominaga, M. Murata, Y. Tanaka, Mol. Ther. Methods Clin. Dev. 3 (2016) 16009. https://doi.org/10.1038/mtm.2016.9
  49. X. Yin, B.E. Mead, H. Safaee, R. Langer, J.M. Karp, O. Levy, Cell Stem Cell 18 (2016) 25. https://doi.org/10.1016/j.stem.2015.12.005
  50. M. Ravi, V. Paramesh, S. Kaviya, E. Anuradha, F. Solomon, J. Cell. Physiol. 230 (2015) 16. https://doi.org/10.1002/jcp.24683
  51. Y. Lei, D.V. Schaffer, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) E5039. https://doi.org/10.1073/pnas.1309408110
  52. G. Turturici, R. Tinnirello, G. Sconzo, F. Geraci, Am. J. Physiol. Cell Physiol. 306 (2014) C621. https://doi.org/10.1152/ajpcell.00228.2013
  53. G. Camussi, M.C. Deregibus, S. Bruno, V. Cantaluppi, L. Biancone, Kidney Int. 78 (2010) 838. https://doi.org/10.1038/ki.2010.278
  54. H.H. Yoon, S.H. Bhang, T. Kim, T. Yu, T. Hyeon, B.S. Kim, Adv. Funct. Mater. 24 (2014) 6455. https://doi.org/10.1002/adfm.201400793
  55. R.A. Rossello, D.H. Kohn, Commun. Integr. Biol. 3 (2010) 53. https://doi.org/10.4161/cib.3.1.9863
  56. D.J. Richards, Y. Tan, R. Coyle, Y. Li, R. Xu, N. Yeung, A. Parker, D.R. Menick, B. Tian, Y. Mei, Nano Lett. 16 (2016) 4670. https://doi.org/10.1021/acs.nanolett.6b02093
  57. R.A. Rossello, Z. Wang, E. Kizana, P.H. Krebsbach, D.H. Kohn, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 13219. https://doi.org/10.1073/pnas.0902622106
  58. C. Frantz, K.M. Stewart, V.M. Weaver, J. Cell Sci. 123 (2010) 4195. https://doi.org/10.1242/jcs.023820
  59. M.W. Bobbie, S. Roy, K. Trudeau, S.J. Munger, A.M. Simon, S. Roy, Investig. Ophthalmol. Vis. Sci. 51 (2010) 3758. https://doi.org/10.1167/iovs.09-4489
  60. S.S. Ho, K.C. Murphy, B.Y. Binder, C.B. Vissers, J.K. Leach, Stem Cells Transl. Med. 5 (2016) 773. https://doi.org/10.5966/sctm.2015-0211
  61. L. Seguin, J.S. Desgrosellier, S.M. Weis, D.A. Cheresh, Trends Cell Biol. 25 (2015) 234. https://doi.org/10.1016/j.tcb.2014.12.006
  62. F.G. Giancotti, E. Ruoslahti, Science 285 (1999) 1028. https://doi.org/10.1126/science.285.5430.1028
  63. T.H. Qazi, D.J. Mooney, G.N. Duda, S. Geissler, Biomaterials (2017).

Cited by

  1. Polydopamine-assisted one-step modification of nanofiber surfaces with adenosine to tune the osteogenic differentiation of mesenchymal stem cells and the maturation of osteoclasts vol.8, pp.10, 2018, https://doi.org/10.1039/c9bm01990a
  2. Engineering Multi‐Cellular Spheroids for Tissue Engineering and Regenerative Medicine vol.9, pp.23, 2018, https://doi.org/10.1002/adhm.202000608
  3. Bioactive Electrospun Fibers: Fabrication Strategies and a Critical Review of Surface-Sensitive Characterization and Quantification vol.121, pp.18, 2018, https://doi.org/10.1021/acs.chemrev.0c00816
  4. Chitin Nerve Conduits with Three-Dimensional Spheroids of Mesenchymal Stem Cells from SD Rats Promote Peripheral Nerve Regeneration vol.13, pp.22, 2018, https://doi.org/10.3390/polym13223957